首页 > 最新文献

Molecular Cancer最新文献

英文 中文
Super-enhancer omics in stem cell 干细胞中的超级增殖物
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-01 DOI: 10.1186/s12943-024-02066-z
Hongying Ma, Jian Qu, Zicheng Pang, Jian Luo, Min Yan, Weixin Xu, Haihui Zhuang, Linxin Liu, Qiang Qu
The hallmarks of stem cells, such as proliferation, self-renewal, development, differentiation, and regeneration, are critical to maintain stem cell identity which is sustained by genetic and epigenetic factors. Super-enhancers (SEs), which consist of clusters of active enhancers, play a central role in maintaining stemness hallmarks by specifically transcriptional model. The SE-navigated transcriptional complex, including SEs, non-coding RNAs, master transcriptional factors, Mediators and other co-activators, forms phase-separated condensates, which offers a toggle for directing diverse stem cell fate. With the burgeoning technologies of multiple-omics applied to examine different aspects of SE, we firstly raise the concept of “super-enhancer omics”, inextricably linking to Pan-omics. In the review, we discuss the spatiotemporal organization and concepts of SEs, and describe links between SE-navigated transcriptional complex and stem cell features, such as stem cell identity, self-renewal, pluripotency, differentiation and development. We also elucidate the mechanism of stemness and oncogenic SEs modulating cancer stem cells via genomic and epigenetic alterations hijack in cancer stem cell. Additionally, we discuss the potential of targeting components of the SE complex using small molecule compounds, genome editing, and antisense oligonucleotides to treat SE-associated organ dysfunction and diseases, including cancer. This review also provides insights into the future of stem cell research through the paradigm of SEs.
干细胞的特征,如增殖、自我更新、发育、分化和再生,是维持干细胞特性的关键,而干细胞特性是由遗传和表观遗传因素维持的。超级增强子(SE)由活性增强子群组成,通过特异性转录模型在维持干性特征方面发挥核心作用。由SE导航的转录复合体,包括SEs、非编码RNAs、主转录因子、Mediators和其他共激活因子,形成相分离的凝聚体,为指导干细胞的多样化命运提供了一个切换器。随着多重组学技术的蓬勃发展,我们首先提出了与泛组学密不可分的 "超级增强子组学 "概念。在综述中,我们讨论了SE的时空组织和概念,描述了SE导航的转录复合物与干细胞特征(如干细胞特性、自我更新、多能性、分化和发育)之间的联系。我们还阐明了干性和致癌SE通过劫持癌症干细胞的基因组和表观遗传学改变来调节癌症干细胞的机制。此外,我们还讨论了利用小分子化合物、基因组编辑和反义寡核苷酸靶向SE复合体成分治疗SE相关器官功能障碍和疾病(包括癌症)的潜力。这篇综述还通过SE的范例为干细胞研究的未来提供了见解。
{"title":"Super-enhancer omics in stem cell","authors":"Hongying Ma, Jian Qu, Zicheng Pang, Jian Luo, Min Yan, Weixin Xu, Haihui Zhuang, Linxin Liu, Qiang Qu","doi":"10.1186/s12943-024-02066-z","DOIUrl":"https://doi.org/10.1186/s12943-024-02066-z","url":null,"abstract":"The hallmarks of stem cells, such as proliferation, self-renewal, development, differentiation, and regeneration, are critical to maintain stem cell identity which is sustained by genetic and epigenetic factors. Super-enhancers (SEs), which consist of clusters of active enhancers, play a central role in maintaining stemness hallmarks by specifically transcriptional model. The SE-navigated transcriptional complex, including SEs, non-coding RNAs, master transcriptional factors, Mediators and other co-activators, forms phase-separated condensates, which offers a toggle for directing diverse stem cell fate. With the burgeoning technologies of multiple-omics applied to examine different aspects of SE, we firstly raise the concept of “super-enhancer omics”, inextricably linking to Pan-omics. In the review, we discuss the spatiotemporal organization and concepts of SEs, and describe links between SE-navigated transcriptional complex and stem cell features, such as stem cell identity, self-renewal, pluripotency, differentiation and development. We also elucidate the mechanism of stemness and oncogenic SEs modulating cancer stem cells via genomic and epigenetic alterations hijack in cancer stem cell. Additionally, we discuss the potential of targeting components of the SE complex using small molecule compounds, genome editing, and antisense oligonucleotides to treat SE-associated organ dysfunction and diseases, including cancer. This review also provides insights into the future of stem cell research through the paradigm of SEs.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141862174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting DKK1 enhances the antitumor activity of paclitaxel and alleviates chemotherapy-induced peripheral neuropathy in breast cancer 靶向 DKK1 可增强紫杉醇的抗肿瘤活性并缓解化疗引起的乳腺癌周围神经病变
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-31 DOI: 10.1186/s12943-024-02067-y
Hong-Xiang Shi, Hang-Tian Tao, Jin-Jin He, Feng-Yi Zhu, Cui-Qing Xie, Yu-Na Cheng, Li-Li Hou, Hua Sun, Chang-Jiang Qin, Dong Fang, Song-Qiang Xie
Chemotherapy in combination with immunotherapy has gradually shown substantial promise to increase T cell infiltration and antitumor efficacy. However, paclitaxel in combination with immune checkpoint inhibitor targeting PD-1/PD-L1 was only used to treat a small proportion of metastatic triple-negative breast cancer (TNBC), and the clinical outcomes was very limited. In addition, this regimen cannot prevent paclitaxel-induced peripheral neuropathy. Therefore, there was an urgent need for a novel target to enhance the antitumor activity of paclitaxel and alleviate chemotherapy-induced peripheral neuropathy in breast cancer. Here, we found that Dickkopf-1 (DKK1) expression was upregulated in multiply subtypes of human breast cancer specimens after paclitaxel-based chemotherapy. Mechanistic studies revealed that paclitaxel promoted DKK1 expression by inducing EGFR signaling in breast cancer cells, and the upregulation of DKK1 could hinder the therapeutic efficacy of paclitaxel by suppressing the infiltration and activity of CD8+ T cells in tumor microenvironment. Moreover, paclitaxel treatment in tumor-bearing mice also increased DKK1 expression through the activation of EGFR signaling in the primary sensory dorsal root ganglion (DRG) neurons, leading to the development of peripheral neuropathy, which is charactered by myelin damage in the sciatic nerve, neuropathic pain, and loss of cutaneous innervation in hindpaw skin. The addition of an anti-DKK1 antibody not only improved therapeutic efficacy of paclitaxel in two murine subtype models of breast cancer but also alleviated paclitaxel-induced peripheral neuropathy. Taken together, our findings providing a potential chemoimmunotherapy strategy with low neurotoxicity that can benefit multiple subtypes of breast cancer patients.
化疗联合免疫疗法已逐渐显示出增加T细胞浸润和抗肿瘤疗效的巨大前景。然而,紫杉醇联合靶向PD-1/PD-L1的免疫检查点抑制剂仅用于治疗一小部分转移性三阴性乳腺癌(TNBC),临床疗效非常有限。此外,这种疗法无法预防紫杉醇诱导的周围神经病变。因此,迫切需要一种新的靶点来增强紫杉醇的抗肿瘤活性,缓解乳腺癌化疗引起的周围神经病变。在这里,我们发现在以紫杉醇为基础的化疗后,Dickkopf-1(DKK1)在多种亚型的人类乳腺癌标本中表达上调。机理研究发现,紫杉醇通过诱导乳腺癌细胞的表皮生长因子受体(EGFR)信号转导促进DKK1的表达,而DKK1的上调会抑制CD8+ T细胞在肿瘤微环境中的浸润和活性,从而阻碍紫杉醇的疗效。此外,紫杉醇治疗肿瘤小鼠也会通过激活初级感觉背根神经节(DRG)神经元的表皮生长因子受体(EGFR)信号转导,增加DKK1的表达,从而导致以坐骨神经髓鞘损伤、神经性疼痛和后爪皮肤神经支配丧失为特征的周围神经病变的发生。添加抗 DKK1 抗体不仅能提高紫杉醇在两种乳腺癌亚型小鼠模型中的疗效,还能减轻紫杉醇诱导的周围神经病变。综上所述,我们的研究结果提供了一种潜在的低神经毒性化疗免疫疗法策略,可造福多种亚型乳腺癌患者。
{"title":"Targeting DKK1 enhances the antitumor activity of paclitaxel and alleviates chemotherapy-induced peripheral neuropathy in breast cancer","authors":"Hong-Xiang Shi, Hang-Tian Tao, Jin-Jin He, Feng-Yi Zhu, Cui-Qing Xie, Yu-Na Cheng, Li-Li Hou, Hua Sun, Chang-Jiang Qin, Dong Fang, Song-Qiang Xie","doi":"10.1186/s12943-024-02067-y","DOIUrl":"https://doi.org/10.1186/s12943-024-02067-y","url":null,"abstract":"Chemotherapy in combination with immunotherapy has gradually shown substantial promise to increase T cell infiltration and antitumor efficacy. However, paclitaxel in combination with immune checkpoint inhibitor targeting PD-1/PD-L1 was only used to treat a small proportion of metastatic triple-negative breast cancer (TNBC), and the clinical outcomes was very limited. In addition, this regimen cannot prevent paclitaxel-induced peripheral neuropathy. Therefore, there was an urgent need for a novel target to enhance the antitumor activity of paclitaxel and alleviate chemotherapy-induced peripheral neuropathy in breast cancer. Here, we found that Dickkopf-1 (DKK1) expression was upregulated in multiply subtypes of human breast cancer specimens after paclitaxel-based chemotherapy. Mechanistic studies revealed that paclitaxel promoted DKK1 expression by inducing EGFR signaling in breast cancer cells, and the upregulation of DKK1 could hinder the therapeutic efficacy of paclitaxel by suppressing the infiltration and activity of CD8+ T cells in tumor microenvironment. Moreover, paclitaxel treatment in tumor-bearing mice also increased DKK1 expression through the activation of EGFR signaling in the primary sensory dorsal root ganglion (DRG) neurons, leading to the development of peripheral neuropathy, which is charactered by myelin damage in the sciatic nerve, neuropathic pain, and loss of cutaneous innervation in hindpaw skin. The addition of an anti-DKK1 antibody not only improved therapeutic efficacy of paclitaxel in two murine subtype models of breast cancer but also alleviated paclitaxel-induced peripheral neuropathy. Taken together, our findings providing a potential chemoimmunotherapy strategy with low neurotoxicity that can benefit multiple subtypes of breast cancer patients.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141857856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Circular RNA hsa_circ_0000467 promotes colorectal cancer progression by promoting eIF4A3-mediated c-Myc translation 环状 RNA hsa_circ_0000467 通过促进 eIF4A3 介导的 c-Myc 翻译来推动结直肠癌的进展
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-31 DOI: 10.1186/s12943-024-02052-5
Xianjie Jiang, Mingjing Peng, Qiang Liu, Qiu Peng, Linda Oyang, Shizhen Li, Xuemeng Xu, Mengzhou Shen, Jiewen Wang, Haofan Li, Nayiyuan Wu, Shiming Tan, Jinguan Lin, Longzheng Xia, Yanyan Tang, Xia Luo, Qianjin Liao, Yujuan Zhou
Colorectal cancer (CRC) is the second most common malignant tumor worldwide, and its incidence rate increases annually. Early diagnosis and treatment are crucial for improving the prognosis of patients with colorectal cancer. Circular RNAs are noncoding RNAs with a closed-loop structure that play a significant role in tumor development. However, the role of circular RNAs in CRC is poorly understood. The circular RNA hsa_circ_0000467 was screened in CRC circRNA microarrays using a bioinformatics analysis, and the expression of hsa_circ_0000467 in CRC tissues was determined by in situ hybridization. The associations between the expression level of hsa_circ_0000467 and the clinical characteristics of CRC patients were evaluated. Then, the role of hsa_circ_0000467 in CRC growth and metastasis was assessed by CCK8 assay, EdU assay, plate colony formation assay, wound healing assay, and Transwell assay in vitro and in a mouse model of CRC in vivo. Proteomic analysis and western blotting were performed to investigate the effect of hsa_circ_0000467 on c-Myc signaling. Polysome profiling, RT‒qPCR and dual-luciferase reporter assays were performed to determine the effect of hsa_circ_0000467 on c-Myc translation. RNA pull-down, RNA immunoprecipitation (RIP) and immunofluorescence staining were performed to assess the effect of hsa_circ_0000467 on eIF4A3 distribution. In this study, we found that the circular RNA hsa_circ_0000467 is highly expressed in colorectal cancer and is significantly correlated with poor prognosis in CRC patients. In vitro and in vivo experiments revealed that hsa_circ_0000467 promotes the growth and metastasis of colorectal cancer cells. Mechanistically, hsa_circ_0000467 binds eIF4A3 to suppress its nuclear translocation. In addition, it can also act as a scaffold molecule that binds eIF4A3 and c-Myc mRNA to form complexes in the cytoplasm, thereby promoting the translation of c-Myc. In turn, c-Myc upregulates its downstream targets, including the cell cycle-related factors cyclin D2 and CDK4 and the tight junction-related factor ZEB1, and downregulates E-cadherin, which ultimately promotes the growth and metastasis of CRC. Our findings revealed that hsa_circRNA_0000467 plays a role in the progression of CRC by promoting eIF4A3-mediated c-Myc translation. This study provides a theoretical basis and molecular target for the diagnosis and treatment of CRC.
结直肠癌(CRC)是全球第二大常见恶性肿瘤,其发病率逐年上升。早期诊断和治疗对于改善结直肠癌患者的预后至关重要。环状 RNA 是具有闭环结构的非编码 RNA,在肿瘤发生发展过程中发挥着重要作用。然而,人们对环状 RNA 在 CRC 中的作用还知之甚少。通过生物信息学分析,在 CRC circRNA 微阵列中筛选出了环状 RNA hsa_circ_0000467,并通过原位杂交测定了 hsa_circ_0000467 在 CRC 组织中的表达。评估了 hsa_circ_0000467 的表达水平与 CRC 患者临床特征之间的关联。然后,通过 CCK8 试验、EdU 试验、平板集落形成试验、伤口愈合试验和 Transwell 试验,在体外和小鼠 CRC 模型中评估了 hsa_circ_0000467 在 CRC 生长和转移中的作用。为了研究 hsa_circ_0000467 对 c-Myc 信号转导的影响,进行了蛋白质组分析和 Western 印迹。为了确定 hsa_circ_0000467 对 c-Myc 翻译的影响,进行了多聚体分析、RT-qPCR 和双荧光素酶报告实验。为了评估 hsa_circ_0000467 对 eIF4A3 分布的影响,我们进行了 RNA 拉取、RNA 免疫沉淀(RIP)和免疫荧光染色。在这项研究中,我们发现环状 RNA hsa_circ_0000467 在结直肠癌中高表达,并与 CRC 患者的不良预后显著相关。体外和体内实验显示,hsa_circ_0000467 能促进结直肠癌细胞的生长和转移。从机理上讲,hsa_circ_0000467 与 eIF4A3 结合,抑制其核转位。此外,它还可以作为支架分子,与 eIF4A3 和 c-Myc mRNA 结合,在细胞质中形成复合物,从而促进 c-Myc 的翻译。反过来,c-Myc 会上调其下游靶标,包括细胞周期相关因子细胞周期蛋白 D2 和 CDK4 以及紧密连接相关因子 ZEB1,并下调 E-cadherin,最终促进 CRC 的生长和转移。我们的研究结果表明,hsa_circRNA_0000467通过促进eIF4A3介导的c-Myc翻译,在CRC的进展过程中发挥作用。这项研究为诊断和治疗 CRC 提供了理论依据和分子靶点。
{"title":"Circular RNA hsa_circ_0000467 promotes colorectal cancer progression by promoting eIF4A3-mediated c-Myc translation","authors":"Xianjie Jiang, Mingjing Peng, Qiang Liu, Qiu Peng, Linda Oyang, Shizhen Li, Xuemeng Xu, Mengzhou Shen, Jiewen Wang, Haofan Li, Nayiyuan Wu, Shiming Tan, Jinguan Lin, Longzheng Xia, Yanyan Tang, Xia Luo, Qianjin Liao, Yujuan Zhou","doi":"10.1186/s12943-024-02052-5","DOIUrl":"https://doi.org/10.1186/s12943-024-02052-5","url":null,"abstract":"Colorectal cancer (CRC) is the second most common malignant tumor worldwide, and its incidence rate increases annually. Early diagnosis and treatment are crucial for improving the prognosis of patients with colorectal cancer. Circular RNAs are noncoding RNAs with a closed-loop structure that play a significant role in tumor development. However, the role of circular RNAs in CRC is poorly understood. The circular RNA hsa_circ_0000467 was screened in CRC circRNA microarrays using a bioinformatics analysis, and the expression of hsa_circ_0000467 in CRC tissues was determined by in situ hybridization. The associations between the expression level of hsa_circ_0000467 and the clinical characteristics of CRC patients were evaluated. Then, the role of hsa_circ_0000467 in CRC growth and metastasis was assessed by CCK8 assay, EdU assay, plate colony formation assay, wound healing assay, and Transwell assay in vitro and in a mouse model of CRC in vivo. Proteomic analysis and western blotting were performed to investigate the effect of hsa_circ_0000467 on c-Myc signaling. Polysome profiling, RT‒qPCR and dual-luciferase reporter assays were performed to determine the effect of hsa_circ_0000467 on c-Myc translation. RNA pull-down, RNA immunoprecipitation (RIP) and immunofluorescence staining were performed to assess the effect of hsa_circ_0000467 on eIF4A3 distribution. In this study, we found that the circular RNA hsa_circ_0000467 is highly expressed in colorectal cancer and is significantly correlated with poor prognosis in CRC patients. In vitro and in vivo experiments revealed that hsa_circ_0000467 promotes the growth and metastasis of colorectal cancer cells. Mechanistically, hsa_circ_0000467 binds eIF4A3 to suppress its nuclear translocation. In addition, it can also act as a scaffold molecule that binds eIF4A3 and c-Myc mRNA to form complexes in the cytoplasm, thereby promoting the translation of c-Myc. In turn, c-Myc upregulates its downstream targets, including the cell cycle-related factors cyclin D2 and CDK4 and the tight junction-related factor ZEB1, and downregulates E-cadherin, which ultimately promotes the growth and metastasis of CRC. Our findings revealed that hsa_circRNA_0000467 plays a role in the progression of CRC by promoting eIF4A3-mediated c-Myc translation. This study provides a theoretical basis and molecular target for the diagnosis and treatment of CRC.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141857792","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Decoding the spatiotemporal heterogeneity of tumor-associated macrophages 解码肿瘤相关巨噬细胞的时空异质性
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-27 DOI: 10.1186/s12943-024-02064-1
Xiangyuan Chu, Yu Tian, Chao Lv
Tumor-associated macrophages (TAMs) are pivotal in cancer progression, influencing tumor growth, angiogenesis, and immune evasion. This review explores the spatial and temporal heterogeneity of TAMs within the tumor microenvironment (TME), highlighting their diverse subtypes, origins, and functions. Advanced technologies such as single-cell sequencing and spatial multi-omics have elucidated the intricate interactions between TAMs and other TME components, revealing the mechanisms behind their recruitment, polarization, and distribution. Key findings demonstrate that TAMs support tumor vascularization, promote epithelial-mesenchymal transition (EMT), and modulate extracellular matrix (ECM) remodeling, etc., thereby enhancing tumor invasiveness and metastasis. Understanding these complex dynamics offers new therapeutic targets for disrupting TAM-mediated pathways and overcoming drug resistance. This review underscores the potential of targeting TAMs to develop innovative cancer therapies, emphasizing the need for further research into their spatial characteristics and functional roles within the TME.
肿瘤相关巨噬细胞(TAMs)在癌症进展中起着关键作用,影响着肿瘤的生长、血管生成和免疫逃避。本综述探讨了 TAMs 在肿瘤微环境(TME)中的时空异质性,重点介绍了它们的不同亚型、起源和功能。单细胞测序和空间多组学等先进技术阐明了TAMs与其他TME成分之间错综复杂的相互作用,揭示了它们招募、极化和分布背后的机制。主要研究结果表明,TAMs 支持肿瘤血管化、促进上皮-间质转化(EMT)、调节细胞外基质(ECM)重塑等,从而增强了肿瘤的侵袭性和转移性。了解这些复杂的动态变化为破坏 TAM 介导的通路和克服耐药性提供了新的治疗靶点。这篇综述强调了靶向 TAMs 开发创新癌症疗法的潜力,同时强调了进一步研究 TAM 在 TME 中的空间特征和功能作用的必要性。
{"title":"Decoding the spatiotemporal heterogeneity of tumor-associated macrophages","authors":"Xiangyuan Chu, Yu Tian, Chao Lv","doi":"10.1186/s12943-024-02064-1","DOIUrl":"https://doi.org/10.1186/s12943-024-02064-1","url":null,"abstract":"Tumor-associated macrophages (TAMs) are pivotal in cancer progression, influencing tumor growth, angiogenesis, and immune evasion. This review explores the spatial and temporal heterogeneity of TAMs within the tumor microenvironment (TME), highlighting their diverse subtypes, origins, and functions. Advanced technologies such as single-cell sequencing and spatial multi-omics have elucidated the intricate interactions between TAMs and other TME components, revealing the mechanisms behind their recruitment, polarization, and distribution. Key findings demonstrate that TAMs support tumor vascularization, promote epithelial-mesenchymal transition (EMT), and modulate extracellular matrix (ECM) remodeling, etc., thereby enhancing tumor invasiveness and metastasis. Understanding these complex dynamics offers new therapeutic targets for disrupting TAM-mediated pathways and overcoming drug resistance. This review underscores the potential of targeting TAMs to develop innovative cancer therapies, emphasizing the need for further research into their spatial characteristics and functional roles within the TME.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141768564","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ubiquitination and deubiquitination in cancer: from mechanisms to novel therapeutic approaches 癌症中的泛素化和去泛素化:从机制到新型治疗方法
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-25 DOI: 10.1186/s12943-024-02046-3
Fangfang Liu, Jingyu Chen, Kai Li, Haochen Li, Yiyi Zhu, Yubo Zhai, Bingbing Lu, Yanle Fan, Ziyue Liu, Xiaojie Chen, Xuechao Jia, Zigang Dong, Kangdong Liu
Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin–proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.
泛素化是蛋白质的一种关键性翻译后修饰,在调节蛋白质稳定性方面发挥着根本性的作用。泛素化和去泛素化酶的失调是各种癌症的共同特征,这凸显了研究泛素连接酶和去泛素化酶(DUBs)以深入了解致癌过程和开发治疗干预措施的必要性。在这篇综述中,我们将讨论泛素-蛋白酶体系统(UPS)在所有癌症特征中的贡献以及药物发现方面的进展。我们深入探讨了 UPS 在肿瘤学中的多种功能,包括它对多种癌症相关通路的调控、它在代谢重编程中的作用、它与肿瘤免疫反应的关系、它在表型可塑性和多态微生物组中的功能以及其他重要的细胞功能。此外,我们还全面概述了利用 UPS 的新型抗癌策略,包括蛋白水解靶向嵌合体 (PROTAC) 和分子胶的开发与应用。
{"title":"Ubiquitination and deubiquitination in cancer: from mechanisms to novel therapeutic approaches","authors":"Fangfang Liu, Jingyu Chen, Kai Li, Haochen Li, Yiyi Zhu, Yubo Zhai, Bingbing Lu, Yanle Fan, Ziyue Liu, Xiaojie Chen, Xuechao Jia, Zigang Dong, Kangdong Liu","doi":"10.1186/s12943-024-02046-3","DOIUrl":"https://doi.org/10.1186/s12943-024-02046-3","url":null,"abstract":"Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin–proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141754720","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Loss of the tumour suppressor LKB1/STK11 uncovers a leptin-mediated sensitivity mechanism to mitochondrial uncouplers for targeted cancer therapy 肿瘤抑制因子 LKB1/STK11 的缺失揭示了瘦素介导的线粒体解偶联剂对癌症靶向治疗的敏感性机制
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-25 DOI: 10.1186/s12943-024-02061-4
Andriani Angelopoulou, Giorgos Theocharous, Dimitrios Valakos, Aikaterini Polyzou, Sophia Magkouta, Vassilios Myrianthopoulos, Sophia Havaki, Marco Fiorillo, Ioanna Tremi, Konstantinos Vachlas, Theodoros Nisotakis, Dimitris-Foivos Thanos, Anastasia Pantazaki, Dimitris Kletsas, Jiri Bartek, Russell Petty, Dimitris Thanos, Rory J McCrimmon, Angelos Papaspyropoulos, Vassilis G Gorgoulis
Non-small cell lung cancer (NSCLC) constitutes one of the deadliest and most common malignancies. The LKB1/STK11 tumour suppressor is mutated in ∼ 30% of NSCLCs, typically lung adenocarcinomas (LUAD). We implemented zebrafish and human lung organoids as synergistic platforms to pre-clinically screen for metabolic compounds selectively targeting LKB1-deficient tumours. Interestingly, two kinase inhibitors, Piceatannol and Tyrphostin 23, appeared to exert synthetic lethality with LKB1 mutations. Although LKB1 loss alone accelerates energy expenditure, unexpectedly we find that it additionally alters regulation of the key energy homeostasis maintenance player leptin (LEP), further increasing the energetic burden and exposing a vulnerable point; acquired sensitivity to the identified compounds. We show that compound treatment stabilises Hypoxia-inducible factor 1-alpha (HIF1A) by antagonising Von Hippel-Lindau (VHL)-mediated HIF1A ubiquitination, driving LEP hyperactivation. Importantly, we demonstrate that sensitivity to piceatannol/tyrphostin 23 epistatically relies on a HIF1A-LEP-Uncoupling Protein 2 (UCP2) signaling axis lowering cellular energy beyond survival, in already challenged LKB1-deficient cells. Thus, we uncover a pivotal metabolic vulnerability of LKB1-deficient tumours, which may be therapeutically exploited using our identified compounds as mitochondrial uncouplers.
非小细胞肺癌(NSCLC)是最致命、最常见的恶性肿瘤之一。LKB1/STK11肿瘤抑制因子在30%的NSCLC(典型的肺腺癌)中发生突变。我们将斑马鱼和人肺器官组织作为协同平台,对选择性靶向LKB1缺陷肿瘤的代谢化合物进行临床前筛选。有趣的是,Piceatannol 和 Tyrphostin 23 这两种激酶抑制剂似乎对 LKB1 突变具有合成致死作用。虽然 LKB1 的缺失本身会加速能量消耗,但我们意外地发现,它还会改变维持能量平衡的关键因子瘦素(LEP)的调节,从而进一步增加能量负担,并暴露出一个薄弱点,即对已确定化合物的后天敏感性。我们的研究表明,化合物治疗通过拮抗 Von Hippel-Lindau (VHL) 介导的 HIF1A 泛素化,稳定了低氧诱导因子 1-α(HIF1A),从而推动了瘦素的过度激活。重要的是,我们证明,在已经受到挑战的 LKB1 基因缺陷细胞中,对 piceatannol/tyrphostin 23 的敏感性在表观上依赖于 HIF1A-LEP-Uncoupling Protein 2 (UCP2) 信号轴,该信号轴降低了细胞的能量,使细胞无法存活。因此,我们发现了缺乏 LKB1 的肿瘤在新陈代谢方面的一个关键弱点,可以利用我们发现的化合物作为线粒体解偶联剂对其进行治疗。
{"title":"Loss of the tumour suppressor LKB1/STK11 uncovers a leptin-mediated sensitivity mechanism to mitochondrial uncouplers for targeted cancer therapy","authors":"Andriani Angelopoulou, Giorgos Theocharous, Dimitrios Valakos, Aikaterini Polyzou, Sophia Magkouta, Vassilios Myrianthopoulos, Sophia Havaki, Marco Fiorillo, Ioanna Tremi, Konstantinos Vachlas, Theodoros Nisotakis, Dimitris-Foivos Thanos, Anastasia Pantazaki, Dimitris Kletsas, Jiri Bartek, Russell Petty, Dimitris Thanos, Rory J McCrimmon, Angelos Papaspyropoulos, Vassilis G Gorgoulis","doi":"10.1186/s12943-024-02061-4","DOIUrl":"https://doi.org/10.1186/s12943-024-02061-4","url":null,"abstract":"Non-small cell lung cancer (NSCLC) constitutes one of the deadliest and most common malignancies. The LKB1/STK11 tumour suppressor is mutated in ∼ 30% of NSCLCs, typically lung adenocarcinomas (LUAD). We implemented zebrafish and human lung organoids as synergistic platforms to pre-clinically screen for metabolic compounds selectively targeting LKB1-deficient tumours. Interestingly, two kinase inhibitors, Piceatannol and Tyrphostin 23, appeared to exert synthetic lethality with LKB1 mutations. Although LKB1 loss alone accelerates energy expenditure, unexpectedly we find that it additionally alters regulation of the key energy homeostasis maintenance player leptin (LEP), further increasing the energetic burden and exposing a vulnerable point; acquired sensitivity to the identified compounds. We show that compound treatment stabilises Hypoxia-inducible factor 1-alpha (HIF1A) by antagonising Von Hippel-Lindau (VHL)-mediated HIF1A ubiquitination, driving LEP hyperactivation. Importantly, we demonstrate that sensitivity to piceatannol/tyrphostin 23 epistatically relies on a HIF1A-LEP-Uncoupling Protein 2 (UCP2) signaling axis lowering cellular energy beyond survival, in already challenged LKB1-deficient cells. Thus, we uncover a pivotal metabolic vulnerability of LKB1-deficient tumours, which may be therapeutically exploited using our identified compounds as mitochondrial uncouplers.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141754704","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Expression and inactivation of glycogen synthase kinase 3 alpha/ beta and their association with the expression of cyclin D1 and p53 in oral squamous cell carcinoma progression 更正:糖原合酶激酶3α/β的表达和失活及其与口腔鳞状细胞癌进展过程中细胞周期蛋白D1和p53表达的关系
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-25 DOI: 10.1186/s12943-024-02058-z
Rajakishore Mishra, Siddavaram Nagini, Ajay Rana

Correction: Mol Cancer 14, 20 (2015)

https://doi.org/10.1186/s12943-015-0300-x


Following publication of the original article [1], the authors learned that an incorrect representative IHC image (Fig. 1n) for GSK3beta was used. We have identified the error and would like to replace it with the corrected Fig. 1n. The image replacement does not alter the paper's conclusions, and our proposed hypothesis remains the same. They apologize for this oversight.

The incorrect Fig. 1:

Fig. 1
figure 1

The expression of GSK3α and GSK3β proteins in the tumor/ normal tissues of various anatomical sites of the mouth. (A) Representative immunostaining showing the differential expression of GSK3α and GSK3β from consecutive sections in various types of oral tumor tissue samples as indicated in the figure. (a, b) SCC (cheek); (c, d) Mucoepidermoid carcinoma (palate); (e, f) Adamantinoma (mandible); (g, h) SCC (gingiva); (i, j) SCC (lower mandible); (k, l) Adenoid cystic carcinoma (palate); (m, n) Basal cell carcinoma (lip); (o, p) Acinic cell carcinoma (parotid gland); (q, r) Mucoepidermoid carcinoma (root of the tongue). (s, t) Normal salivary gland, (u, v) Mucoepidermoid carcinoma (parotid gland) (w, x) and SCC (lip) showed differential expression of GSK3α and GSK3β. The maximum intense immunoreactivity of GSK3β was observed in SCC compared to other types of oral tumors. Original magnification 100X. (B) Overexpression of GSK3β is significantly higher in SCC than in the other types of (non-SCC) oral cancers (p < 0.0001)

Full size image

The correct Fig. 1:

Fig. 1
figure 2

The expression of GSK3α and GSK3β proteins in the tumor/ normal tissues of various anatomical sites of the mouth. (A) Representative immunostain

转载与授权引用本文Mishra,R.,Nagini,S. &amp; Rana,A. Correction:糖原合酶激酶3α/β的表达和失活及其与口腔鳞状细胞癌进展中细胞周期蛋白D1和p53表达的关系。Mol Cancer 23, 149 (2024). https://doi.org/10.1186/s12943-024-02058-zDownload citationPublished: 25 July 2024DOI: https://doi.org/10.1186/s12943-024-02058-zShare this articleAnyone you share the following link with will be able to read this content:Get shareable linkSorry, a shareable link is not currently available for this article.Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative.
{"title":"Correction: Expression and inactivation of glycogen synthase kinase 3 alpha/ beta and their association with the expression of cyclin D1 and p53 in oral squamous cell carcinoma progression","authors":"Rajakishore Mishra, Siddavaram Nagini, Ajay Rana","doi":"10.1186/s12943-024-02058-z","DOIUrl":"https://doi.org/10.1186/s12943-024-02058-z","url":null,"abstract":"<p><b>Correction: Mol Cancer 14, 20 (2015)</b></p><p><b>https://doi.org/10.1186/s12943-015-0300-x</b></p><br/><p>Following publication of the original article [1], the authors learned that an incorrect representative IHC image (Fig. 1n) for GSK3beta was used. We have identified the error and would like to replace it with the corrected Fig. 1n. The image replacement does not alter the paper's conclusions, and our proposed hypothesis remains the same. They apologize for this oversight.\u0000</p><p>The incorrect Fig. 1:</p><figure><figcaption><b data-test=\"figure-caption-text\">Fig. 1</b></figcaption><picture><source srcset=\"//media.springernature.com/lw685/springer-static/image/art%3A10.1186%2Fs12943-024-02058-z/MediaObjects/12943_2024_2058_Fig1_HTML.png?as=webp\" type=\"image/webp\"/><img alt=\"figure 1\" aria-describedby=\"Fig1\" height=\"1020\" loading=\"lazy\" src=\"//media.springernature.com/lw685/springer-static/image/art%3A10.1186%2Fs12943-024-02058-z/MediaObjects/12943_2024_2058_Fig1_HTML.png\" width=\"685\"/></picture><p><b>The expression of GSK3</b>α <b>and GSK3β</b> <b>proteins in the tumor/ normal tissues of various anatomical sites of the mouth. (A)</b> Representative immunostaining showing the differential expression of GSK3α and GSK3β from consecutive sections in various types of oral tumor tissue samples as indicated in the figure. (a, b) SCC (cheek); (c, d) Mucoepidermoid carcinoma (palate); (e, f) Adamantinoma (mandible); (g, h) SCC (gingiva); (i, j) SCC (lower mandible); (k, l) Adenoid cystic carcinoma (palate); (m, n) Basal cell carcinoma (lip); (o, p) Acinic cell carcinoma (parotid gland); (q, r) Mucoepidermoid carcinoma (root of the tongue). (s, t) Normal salivary gland, (u, v) Mucoepidermoid carcinoma (parotid gland) (w, x) and SCC (lip) showed differential expression of GSK3α and GSK3β. The maximum intense immunoreactivity of GSK3β was observed in SCC compared to other types of oral tumors. Original magnification 100X. <b>(B)</b> Overexpression of GSK3β is significantly higher in SCC than in the other types of (non-SCC) oral cancers (p &lt; 0.0001)</p><span>Full size image</span><svg aria-hidden=\"true\" focusable=\"false\" height=\"16\" role=\"img\" width=\"16\"><use xlink:href=\"#icon-eds-i-chevron-right-small\" xmlns:xlink=\"http://www.w3.org/1999/xlink\"></use></svg></figure><p>The correct Fig. 1:</p><figure><figcaption><b data-test=\"figure-caption-text\">Fig. 1</b></figcaption><picture><source srcset=\"//media.springernature.com/lw685/springer-static/image/art%3A10.1186%2Fs12943-024-02058-z/MediaObjects/12943_2024_2058_Fig2_HTML.png?as=webp\" type=\"image/webp\"/><img alt=\"figure 2\" aria-describedby=\"Fig2\" height=\"1010\" loading=\"lazy\" src=\"//media.springernature.com/lw685/springer-static/image/art%3A10.1186%2Fs12943-024-02058-z/MediaObjects/12943_2024_2058_Fig2_HTML.png\" width=\"685\"/></picture><p><b>The expression of GSK3</b>α <b>and GSK3β</b> <b>proteins in the tumor/ normal tissues of various anatomical sites of the mouth. (A)</b> Representative immunostain","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141754708","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Clinical application of liquid biopsy in colorectal cancer: detection, prediction, and treatment monitoring 液体活检在结直肠癌中的临床应用:检测、预测和治疗监测
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-16 DOI: 10.1186/s12943-024-02063-2
Xiang-Yuan Tao, Qian-Qian Li, Yong Zeng
Colorectal cancer (CRC) is one of the most prevalent malignancies affecting the gastrointestinal tract and is ranked third among cancers with the highest incidence and second-highest mortality rate worldwide. CRC exhibits a slow progression providing a wide treatment window. The currently employed CRC screening methods have shown great potential to prevent CRC and reduce CRC-related morbidity and mortality. The diagnosis of CRC is achieved by colonoscopy and tissue biopsy, with studies showing that liquid biopsy is more effective in detecting and diagnosing early CRC patients. Increasing number of studies have shown that the tumor components shed into circulating blood can be detected in liquid form, and can be applied in the clinical management of CRC. Analysis of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), or tumor-associated platelets (TEPs) in the blood can be used for early screening and diagnosis of CRC, aid tumor staging, treatment response monitoring, and prediction of CRC recurrence and metastasis in a minimally invasive manner. This chapter provides an updated review of CTCs, ctDNA, and TEPs as novel biomarkers for CRC, highlighting their strengths and limitations.
结肠直肠癌(CRC)是影响胃肠道的最常见恶性肿瘤之一,在全球发病率最高、死亡率第二高的癌症中排名第三。CRC 病程进展缓慢,治疗窗口宽广。目前采用的 CRC 筛查方法在预防 CRC 和降低与 CRC 相关的发病率和死亡率方面显示出巨大的潜力。CRC 的诊断通过结肠镜检查和组织活检实现,研究表明液体活检在检测和诊断早期 CRC 患者方面更为有效。越来越多的研究表明,脱落到循环血液中的肿瘤成分可以通过液体形式检测出来,并可应用于 CRC 的临床治疗。对血液中的循环肿瘤细胞(CTCs)、循环肿瘤 DNA(ctDNA)或肿瘤相关血小板(TEPs)进行分析,可用于早期筛查和诊断 CRC,辅助肿瘤分期、治疗反应监测,并以微创方式预测 CRC 的复发和转移。本章对作为 CRC 新型生物标记物的 CTCs、ctDNA 和 TEPs 进行了最新综述,重点介绍了它们的优势和局限性。
{"title":"Clinical application of liquid biopsy in colorectal cancer: detection, prediction, and treatment monitoring","authors":"Xiang-Yuan Tao, Qian-Qian Li, Yong Zeng","doi":"10.1186/s12943-024-02063-2","DOIUrl":"https://doi.org/10.1186/s12943-024-02063-2","url":null,"abstract":"Colorectal cancer (CRC) is one of the most prevalent malignancies affecting the gastrointestinal tract and is ranked third among cancers with the highest incidence and second-highest mortality rate worldwide. CRC exhibits a slow progression providing a wide treatment window. The currently employed CRC screening methods have shown great potential to prevent CRC and reduce CRC-related morbidity and mortality. The diagnosis of CRC is achieved by colonoscopy and tissue biopsy, with studies showing that liquid biopsy is more effective in detecting and diagnosing early CRC patients. Increasing number of studies have shown that the tumor components shed into circulating blood can be detected in liquid form, and can be applied in the clinical management of CRC. Analysis of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), or tumor-associated platelets (TEPs) in the blood can be used for early screening and diagnosis of CRC, aid tumor staging, treatment response monitoring, and prediction of CRC recurrence and metastasis in a minimally invasive manner. This chapter provides an updated review of CTCs, ctDNA, and TEPs as novel biomarkers for CRC, highlighting their strengths and limitations.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141624800","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PD1/PD-L1 blockade in clear cell renal cell carcinoma: mechanistic insights, clinical efficacy, and future perspectives 透明细胞肾细胞癌的 PD1/PD-L1 阻断疗法:机理认识、临床疗效和未来展望
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-16 DOI: 10.1186/s12943-024-02059-y
Zhaoyang Zhu, Yigang Jin, Jing Zhou, Fei Chen, Minjie Chen, Zhaofeng Gao, Lingyu Hu, Jinyan Xuan, Xiaoping Li, Zhengwei Song, Xiao Guo
The advent of PD1/PD-L1 inhibitors has significantly transformed the therapeutic landscape for clear cell renal cell carcinoma (ccRCC). This review provides an in-depth analysis of the biological functions and regulatory mechanisms of PD1 and PD-L1 in ccRCC, emphasizing their role in tumor immune evasion. We comprehensively evaluate the clinical efficacy and safety profiles of PD1/PD-L1 inhibitors, such as Nivolumab and Pembrolizumab, through a critical examination of recent clinical trial data. Furthermore, we discuss the challenges posed by resistance mechanisms to these therapies and potential strategies to overcome them. We also explores the synergistic potential of combination therapies, integrating PD1/PD-L1 inhibitors with other immunotherapies, targeted therapies, and conventional modalities such as chemotherapy and radiotherapy. In addition, we examine emerging predictive biomarkers for response to PD1/PD-L1 blockade and biomarkers indicative of resistance, providing a foundation for personalized therapeutic approaches. Finally, we outline future research directions, highlighting the need for novel therapeutic strategies, deeper mechanistic insights, and the development of individualized treatment regimens. Our work summarizes the latest knowledge and progress in this field, aiming to provide a valuable reference for improving clinical efficacy and guiding future research on the application of PD1/PD-L1 inhibitors in ccRCC.
PD1/PD-L1抑制剂的出现极大地改变了透明细胞肾细胞癌(ccRCC)的治疗格局。本综述深入分析了 PD1 和 PD-L1 在 ccRCC 中的生物学功能和调控机制,强调了它们在肿瘤免疫逃避中的作用。我们通过对近期临床试验数据的严格审查,全面评估了 PD1/PD-L1 抑制剂(如 Nivolumab 和 Pembrolizumab)的临床疗效和安全性。此外,我们还讨论了这些疗法的耐药机制所带来的挑战以及克服这些挑战的潜在策略。我们还探讨了将 PD1/PD-L1 抑制剂与其他免疫疗法、靶向疗法以及化疗和放疗等传统模式相结合的联合疗法的协同潜力。此外,我们还研究了新出现的 PD1/PD-L1 阻断反应的预测性生物标志物和耐药性的指示性生物标志物,为个性化治疗方法奠定了基础。最后,我们概述了未来的研究方向,强调了对新型治疗策略、更深入的机理研究和个体化治疗方案开发的需求。我们的研究总结了这一领域的最新知识和进展,旨在为提高临床疗效提供有价值的参考,并指导未来将 PD1/PD-L1 抑制剂应用于 ccRCC 的研究。
{"title":"PD1/PD-L1 blockade in clear cell renal cell carcinoma: mechanistic insights, clinical efficacy, and future perspectives","authors":"Zhaoyang Zhu, Yigang Jin, Jing Zhou, Fei Chen, Minjie Chen, Zhaofeng Gao, Lingyu Hu, Jinyan Xuan, Xiaoping Li, Zhengwei Song, Xiao Guo","doi":"10.1186/s12943-024-02059-y","DOIUrl":"https://doi.org/10.1186/s12943-024-02059-y","url":null,"abstract":"The advent of PD1/PD-L1 inhibitors has significantly transformed the therapeutic landscape for clear cell renal cell carcinoma (ccRCC). This review provides an in-depth analysis of the biological functions and regulatory mechanisms of PD1 and PD-L1 in ccRCC, emphasizing their role in tumor immune evasion. We comprehensively evaluate the clinical efficacy and safety profiles of PD1/PD-L1 inhibitors, such as Nivolumab and Pembrolizumab, through a critical examination of recent clinical trial data. Furthermore, we discuss the challenges posed by resistance mechanisms to these therapies and potential strategies to overcome them. We also explores the synergistic potential of combination therapies, integrating PD1/PD-L1 inhibitors with other immunotherapies, targeted therapies, and conventional modalities such as chemotherapy and radiotherapy. In addition, we examine emerging predictive biomarkers for response to PD1/PD-L1 blockade and biomarkers indicative of resistance, providing a foundation for personalized therapeutic approaches. Finally, we outline future research directions, highlighting the need for novel therapeutic strategies, deeper mechanistic insights, and the development of individualized treatment regimens. Our work summarizes the latest knowledge and progress in this field, aiming to provide a valuable reference for improving clinical efficacy and guiding future research on the application of PD1/PD-L1 inhibitors in ccRCC.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141624799","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unveiling the PDK4-centered rituximab-resistant mechanism in DLBCL: the potential of the "Smart" exosome nanoparticle therapy. 揭示DLBCL中以PDK4为中心的利妥昔单抗耐药机制:"智能 "外泌体纳米颗粒疗法的潜力。
IF 27.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-15 DOI: 10.1186/s12943-024-02057-0
Xin Wu, Chunmei Ban, Woding Deng, Xuewei Bao, Ning Tang, Yupeng Wu, Zhixuan Deng, Jianbin Xiong, Qiangqiang Zhao

Background: Diffuse large B-cell lymphoma (DLBCL) represents a prevalent malignant tumor, with approximately 40% of patients encountering treatment challenges or relapse attributed to rituximab resistance, primarily due to diminished or absent CD20 expression. Our prior research identified PDK4 as a key driver of rituximab resistance through its negative regulation of CD20 expression. Further investigation into PDK4's resistance mechanism and the development of advanced exosome nanoparticle complexes may unveil novel resistance targets and pave the way for innovative, effective treatment modalities for DLBCL.

Methods: We utilized a DLBCL-resistant cell line with high PDK4 expression (SU-DHL-2/R). We infected it with short hairpin RNA (shRNA) lentivirus for RNA sequencing, aiming to identify significantly downregulated mRNA in resistant cells. Techniques including immunofluorescence, immunohistochemistry, and Western blotting were employed to determine PDK4's localization and expression in resistant cells and its regulatory role in phosphorylation of Histone deacetylase 8 (HDAC8). Furthermore, we engineered advanced exosome nanoparticle complexes, aCD20@ExoCTX/siPDK4, through cellular, genetic, and chemical engineering methods. These nanoparticles underwent characterization via Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM), and their cellular uptake was assessed through flow cytometry. We evaluated the nanoparticles' effects on apoptosis in DLBCL-resistant cells and immune cells using CCK-8 assays and flow cytometry. Additionally, their capacity to counteract resistance and exert anti-tumor effects was tested in a resistant DLBCL mouse model.

Results: We found that PDK4 initiates HDAC8 activation by phosphorylating the Ser-39 site, suppressing CD20 protein expression through deacetylation. The aCD20@ExoCTX/siPDK4 nanoparticles served as effective intracellular delivery mechanisms for gene therapy and monoclonal antibodies, simultaneously inducing apoptosis in resistant DLBCL cells and triggering immunogenic cell death in tumor cells. This dual action effectively reversed the immunosuppressive tumor microenvironment, showcasing a synergistic therapeutic effect in a subcutaneous mouse tumor resistance model.

Conclusions: This study demonstrates that PDK4 contributes to rituximab resistance in DLBCL by modulating CD20 expression via HDAC8 phosphorylation. The designed exosome nanoparticles effectively overcome this resistance by targeting the PDK4/HDAC8/CD20 pathway, representing a promising approach for drug delivery and treating patients with Rituximab-resistant DLBCL.

背景:弥漫大 B 细胞淋巴瘤(DLBCL弥漫性大 B 细胞淋巴瘤(DLBCL)是一种常见的恶性肿瘤,约 40% 的患者因利妥昔单抗耐药而面临治疗难题或复发,主要原因是 CD20 表达减少或缺失。我们之前的研究发现,PDK4 通过负调控 CD20 的表达,是导致利妥昔单抗耐药的关键因素。进一步研究PDK4的耐药机制和开发先进的外泌体纳米颗粒复合物可能会发现新的耐药靶点,并为DLBCL创新、有效的治疗模式铺平道路:我们利用了PDK4高表达的DLBCL耐药细胞系(SU-DHL-2/R)。我们用短发夹RNA(shRNA)慢病毒感染该细胞系,进行RNA测序,旨在确定耐药细胞中显著下调的mRNA。我们采用了免疫荧光、免疫组织化学和 Western 印迹等技术来确定 PDK4 在耐药细胞中的定位和表达及其在组蛋白去乙酰化酶 8 (HDAC8) 磷酸化中的调控作用。此外,我们还通过细胞、遗传和化学工程方法设计了先进的外泌体纳米颗粒复合物--aCD20@ExoCTX/siPDK4。我们通过动态光散射(DLS)和透射电子显微镜(TEM)对这些纳米颗粒进行了表征,并通过流式细胞术评估了它们的细胞摄取情况。我们使用 CCK-8 检测法和流式细胞术评估了纳米颗粒对 DLBCL 抗性细胞和免疫细胞凋亡的影响。此外,我们还在抗药性 DLBCL 小鼠模型中测试了纳米颗粒对抗抗药性和发挥抗肿瘤作用的能力:结果:我们发现PDK4通过磷酸化Ser-39位点启动HDAC8活化,通过去乙酰化抑制CD20蛋白的表达。aCD20@ExoCTX/siPDK4纳米颗粒可作为基因治疗和单克隆抗体的有效细胞内递送机制,同时诱导耐药DLBCL细胞凋亡并引发肿瘤细胞的免疫原性细胞死亡。这种双重作用有效地逆转了免疫抑制性肿瘤微环境,在小鼠皮下肿瘤耐药模型中展示了协同治疗效果:本研究表明,PDK4通过HDAC8磷酸化调节CD20的表达,从而导致DLBCL对利妥昔单抗产生耐药性。所设计的外泌体纳米粒子通过靶向PDK4/HDAC8/CD20通路有效克服了这种耐药性,是治疗利妥昔单抗耐药DLBCL患者的一种很有前景的给药方法。
{"title":"Unveiling the PDK4-centered rituximab-resistant mechanism in DLBCL: the potential of the \"Smart\" exosome nanoparticle therapy.","authors":"Xin Wu, Chunmei Ban, Woding Deng, Xuewei Bao, Ning Tang, Yupeng Wu, Zhixuan Deng, Jianbin Xiong, Qiangqiang Zhao","doi":"10.1186/s12943-024-02057-0","DOIUrl":"10.1186/s12943-024-02057-0","url":null,"abstract":"<p><strong>Background: </strong>Diffuse large B-cell lymphoma (DLBCL) represents a prevalent malignant tumor, with approximately 40% of patients encountering treatment challenges or relapse attributed to rituximab resistance, primarily due to diminished or absent CD20 expression. Our prior research identified PDK4 as a key driver of rituximab resistance through its negative regulation of CD20 expression. Further investigation into PDK4's resistance mechanism and the development of advanced exosome nanoparticle complexes may unveil novel resistance targets and pave the way for innovative, effective treatment modalities for DLBCL.</p><p><strong>Methods: </strong>We utilized a DLBCL-resistant cell line with high PDK4 expression (SU-DHL-2/R). We infected it with short hairpin RNA (shRNA) lentivirus for RNA sequencing, aiming to identify significantly downregulated mRNA in resistant cells. Techniques including immunofluorescence, immunohistochemistry, and Western blotting were employed to determine PDK4's localization and expression in resistant cells and its regulatory role in phosphorylation of Histone deacetylase 8 (HDAC8). Furthermore, we engineered advanced exosome nanoparticle complexes, aCD20@Exo<sup>CTX</sup>/siPDK4, through cellular, genetic, and chemical engineering methods. These nanoparticles underwent characterization via Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM), and their cellular uptake was assessed through flow cytometry. We evaluated the nanoparticles' effects on apoptosis in DLBCL-resistant cells and immune cells using CCK-8 assays and flow cytometry. Additionally, their capacity to counteract resistance and exert anti-tumor effects was tested in a resistant DLBCL mouse model.</p><p><strong>Results: </strong>We found that PDK4 initiates HDAC8 activation by phosphorylating the Ser-39 site, suppressing CD20 protein expression through deacetylation. The aCD20@Exo<sup>CTX</sup>/siPDK4 nanoparticles served as effective intracellular delivery mechanisms for gene therapy and monoclonal antibodies, simultaneously inducing apoptosis in resistant DLBCL cells and triggering immunogenic cell death in tumor cells. This dual action effectively reversed the immunosuppressive tumor microenvironment, showcasing a synergistic therapeutic effect in a subcutaneous mouse tumor resistance model.</p><p><strong>Conclusions: </strong>This study demonstrates that PDK4 contributes to rituximab resistance in DLBCL by modulating CD20 expression via HDAC8 phosphorylation. The designed exosome nanoparticles effectively overcome this resistance by targeting the PDK4/HDAC8/CD20 pathway, representing a promising approach for drug delivery and treating patients with Rituximab-resistant DLBCL.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":27.7,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247735/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141616875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1